Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Genes Dev ; 29(3): 308-21, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644605

ABSTRACT

PR domain-containing 16 (PRDM16) induces expression of brown fat-specific genes in brown and beige adipocytes, although the underlying transcription-related mechanisms remain largely unknown. Here, in vitro studies show that PRDM16, through its zinc finger domains, directly interacts with the MED1 subunit of the Mediator complex, is recruited to the enhancer of the brown fat-specific uncoupling protein 1 (Ucp1) gene through this interaction, and enhances thyroid hormone receptor (TR)-driven transcription in a biochemically defined system in a Mediator-dependent manner, thus providing a direct link to the general transcription machinery. Complementary cell-based studies show that upon forskolin treatment, PRDM16 induces Ucp1 expression in undifferentiated murine embryonic fibroblasts, that this induction depends on MED1 and TR, and, consistent with a direct effect, that PRDM16 is recruited to the Ucp1 enhancer. Related studies have defined MED1 and PRDM16 interaction domains important for Ucp1 versus Ppargc1a induction by PRDM16. These results reveal novel mechanisms for PRDM16 function through the Mediator complex.


Subject(s)
Adipocytes, Brown/cytology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Ion Channels/genetics , Mediator Complex Subunit 1/metabolism , Mitochondrial Proteins/genetics , Transcription Factors/metabolism , Adipocytes, Brown/metabolism , Animals , Cell Line , Colforsin/pharmacology , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental/drug effects , HEK293 Cells , Humans , Mice , Protein Binding , Protein Structure, Tertiary/genetics , Uncoupling Protein 1
2.
Genet Med ; 23(6): 1050-1057, 2021 06.
Article in English | MEDLINE | ID: mdl-33495529

ABSTRACT

PURPOSE: To expand the recent description of a new neurodevelopmental syndrome related to alterations in CDK19. METHODS: Individuals were identified through international collaboration. Functional studies included autophosphorylation assays for CDK19 Gly28Arg and Tyr32His variants and in vivo zebrafish assays of the CDK19G28R and CDK19Y32H. RESULTS: We describe 11 unrelated individuals (age range: 9 months to 14 years) with de novo missense variants mapped to the kinase domain of CDK19, including two recurrent changes at residues Tyr32 and Gly28. In vitro autophosphorylation and substrate phosphorylation assays revealed that kinase activity of protein was lower for p.Gly28Arg and higher for p.Tyr32His substitutions compared with that of the wild-type protein. Injection of CDK19 messenger RNA (mRNA) with either the Tyr32His or the Gly28Arg variants using in vivo zebrafish model significantly increased fraction of embryos with morphological abnormalities. Overall, the phenotype of the now 14 individuals with CDK19-related disorder includes universal developmental delay and facial dysmorphism, hypotonia (79%), seizures (64%), ophthalmologic anomalies (64%), and autism/autistic traits (56%). CONCLUSION: CDK19 de novo missense variants are responsible for a novel neurodevelopmental disorder. Both kinase assay and zebrafish experiments showed that the pathogenetic mechanism may be more diverse than previously thought.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Animals , Cyclin-Dependent Kinases/genetics , Gain of Function Mutation , Humans , Infant , Mutation, Missense , Zebrafish/genetics
3.
Genes Cells ; 23(10): 808-821, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30092612

ABSTRACT

Regulation of the expression of diverse genes is essential for making possible the complexity of higher organisms, and the temporal and spatial regulation of gene expression allows for the alteration of cell types and growth patterns. A critical component of this regulation is the DNA sequence-specific binding of transcription factors (TFs). However, most TFs do not independently participate in gene transcriptional regulation, because they lack an effector function. Instead, TFs are thought to work by recruiting cofactors, including Mediator complex (Mediator), chromatin-remodeling complexes (CRCs), and histone-modifying complexes (HMCs). Mediator associates with the majority of transcribed genes and acts as an integrator of multiple signals. On the other hand, CRCs and HMCs are selectively recruited by TFs. Although all the pairings between TFs and CRCs or HMCs are not fully known, there are a growing number of established TF-CRC and TF-HMC combinations. In this review, we focused on the most important of these pairings and discuss how they control gene expression.


Subject(s)
Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Transcription Factors/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , DNA-Binding Proteins/physiology , Enhancer Elements, Genetic/genetics , Histone Code/genetics , Histones/metabolism , Humans , Mediator Complex/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics
4.
Genes Cells ; 22(3): 265-276, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28151579

ABSTRACT

In eukaryotes, the Mediator complex has important roles in regulation of transcription by RNA polymerase II. Mediator is a large complex with more than 20 subunits that form head, middle, tail and CDK/cyclin modules. Among them, CDK8 and/or CDK19 (CDK8/19), and their counterpart cyclin C, form the CDK/cyclin module together with Mediator subunits MED12 and MED13. Despite evidences of both activation and repression, the precise functional roles of CDK8/19 in transcription are still elusive. Our previous results indicate that CDK8/19 recruits epigenetic regulators to repress immunoresponse genes. Here, this study focused on Toll-like receptors (TLRs), which exert innate immune responses through recognition of pathogen-associated molecular patterns and examined the functional roles of CDK8/19. As a result, CDK8/19 regulated transcription of inflammatory genes on stimulation of TLR9 in myeloma-derived RPMI8226 cells, which led to expression of inflammation-associated genes such as IL8, IL10, PTX3 and CCL2. Mediator subunits CDK8/19 and MED1, inflammation-related transcriptional activator NF-κB and C/EBPß, and general transcription factors TFIIE and TFIIB colocalized at the promoter regions of these genes under this condition. Our results show that CDK8/19 positively regulates inflammatory gene transcription in cooperation with NF-κB and C/EBPß on stimulation of TLR9.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Cyclin-Dependent Kinase 8/physiology , Cyclin-Dependent Kinases/physiology , NF-kappa B/metabolism , Toll-Like Receptor 9/physiology , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Transcriptional Activation
5.
Genes Cells ; 20(3): 203-16, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25492609

ABSTRACT

In eukaryotes, the general transcription factor TFIIE consists of two subunits, α and ß, and plays essential roles in transcription. Structure-function studies indicate that TFIIE has three-winged helix (WH) motifs, with one in TFIIEα and two in TFIIEß. Recent studies suggested that, by binding to the clamp region of RNA polymerase II, TFIIEα-WH promotes the conformational change that transforms the promoter-bound inactive preinitiation complex to the active complex. Here, to elucidate its roles in transcription, functional analyses of point-mutated human TFIIEα-WH proteins were carried out. In vitro transcription analyses identified two classes of mutants. One class was defective in transcription initiation, and the other was defective in the transition from initiation to elongation. Analyses of the binding of this motif to other general transcription factors showed that the former class was defective in binding to the basic helix-loop-helix motif of TFIIEß and the latter class was defective in binding to the N-terminal cyclin homology region of TFIIB. Furthermore, TFIIEα-WH bound to the TFIIH XPB subunit at a third distinct region. Therefore, these results provide further insights into the mechanisms underlying RNA polymerase II activation at the initial stages of transcription.


Subject(s)
Transcription Elongation, Genetic , Transcription Factor TFIIB/metabolism , Transcription Factors, TFII/metabolism , Transcription Initiation, Genetic , Winged-Helix Transcription Factors/metabolism , Animals , CHO Cells , Caenorhabditis elegans , Cricetulus , Drosophila melanogaster , Helix-Loop-Helix Motifs , Humans , Mutation , Saccharomyces cerevisiae , Schizosaccharomyces , Sulfolobus solfataricus , Transcription Factor TFIIH/metabolism , Xenopus laevis
6.
Genes Cells ; 20(3): 191-202, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25482373

ABSTRACT

In eukaryotes, holo-Mediator consists of four modules: head, middle, tail, and CDK/Cyclin. The head module performs an essential function involved in regulation of RNA polymerase II (Pol II). We studied the human head module subunit MED17 (hMED17). Recent structural studies showed that yeast MED17 may function as a hinge connecting the neck and movable jaw regions of the head module to the fixed jaw region. Luciferase assays in hMED17-knockdown cells showed that hMED17 supports transcriptional activation, and pulldown assays showed that hMED17 interacted with Pol II and the general transcription factors TFIIB, TBP, TFIIE, and TFIIH. In addition, hMED17 bound to a DNA helicase subunit of TFIIH, XPB, which is essential for both transcription and nucleotide excision repair (NER). Because hMED17 associates with p53 upon UV-C irradiation, we treated human MCF-7 cells with either UV-C or the MDM2 inhibitor Nutlin-3. Both treatments resulted in accumulation of p53 in the nucleus, but hMED17 remained concentrated in the nucleus in response to UV-C. hMED17 colocalized with the NER factors XPB and XPG following UV-C irradiation, and XPG and XPB bound to hMED17 in vitro. These findings suggest that hMED17 may play essential roles in switching between transcription and NER.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Mediator Complex/metabolism , Transcription Factors/metabolism , Enzyme Inhibitors/pharmacology , HeLa Cells/radiation effects , Humans , Imidazoles/pharmacology , MCF-7 Cells/drug effects , MCF-7 Cells/radiation effects , Mediator Complex/genetics , Piperazines/pharmacology , Protein Binding , Protein Transport/radiation effects , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Polymerase II/metabolism , Transcriptional Activation , Ultraviolet Rays
7.
Genes Cells ; 19(7): 582-93, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24840924

ABSTRACT

The Mediator complex (Mediator) is conserved among eukaryotes and is comprised of head, middle, tail and CDK/cyclin modules. The head module has received the most attention because its interaction with RNA polymerase II (Pol II) and the general transcription factors TFIIH and TBP facilitates phosphorylation of the carboxy-terminal domain (CTD) of the largest subunit of Pol II. We studied the human head module subunit hMED18 to elucidate how Mediator is involved in both transcriptional activation and repression. siRNA-mediated hMED18 depletion augmented transcription, indicating that hMED18 functions in transcriptional repression. Treatment of cells with two histone deacetylase (HDAC) inhibitors, the HDAC inhibitor trichostatin A (TSA) and the SIRT inhibitor nicotinamide showed that this repression was not caused by those HDAC activities. A screen for hMED18-target genes showed that the promoters for cap RNA methyltransferase RNMT-activating mini protein (RAM/FAM103A1) and divalent metal transporter 1 (DMT1/SLC11A2) genes were bound by hMED18. Depletion of hMED18 showed hMED18 and the middle module subunit hMED1 were lost from the promoters of those genes, whereas the CDK/cyclin module subunit hCDK8 remained bound. This indicates a novel transcriptional repression mechanism of hMED18 mediated by hCDK8 and further a novel positive role of free CDK/cyclin module in transcriptional activation. [Correction added on 12 June 2014, after first online publication: SLC11A2 amended from SCL11A2.].


Subject(s)
Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Gene Expression Regulation , HeLa Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Niacinamide/pharmacology , Promoter Regions, Genetic , Protein Subunits/genetics , Protein Subunits/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics
8.
Genes Cells ; 19(12): 879-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25308091

ABSTRACT

In eukaryotes, positive cofactor 4 (PC4) stimulates activator-dependent transcription by facilitating transcription initiation and the transition from initiation to elongation. It also forms homodimers and binds to single-stranded DNA and various transcriptional activators, including the general transcription factor TFIIH. In this study, we further investigated PC4 from Homo sapiens and the nematode Caenorhabditis elegans (hPC4 and cePC4, respectively). hPC4 strongly stimulated transcription on a linearized template, whereas it alleviated transcription on a supercoiled template. Transcriptional stimulation by PC4 was also alleviated by increasing the amount of TFIID. GST pull-down studies with general transcription factors indicated that both hPC4 and cePC4 bind strongly to TFIIB, TFIIEß, TFIIFα, TFIIFß and TFIIH XPB subunits and weakly to TBP and TFIIH p62. However, only hPC4 bound to CDK7. The effect of each PC4 on transcription was studied in combination with TFIIEß. hPC4 stimulated both basal and activated transcription, whereas cePC4 primarily stimulated activated transcription, especially in the presence of TFIIEß from C. elegans. Finally, hPC4 bound to the C-terminal region of hTFIIEß adjacent to the basic region. These results indicate that PC4 plays essential roles in the transition step from transcription initiation to elongation by binding to melted DNA in collaboration with TFIIEß.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors, TFII/metabolism , Transcription Factors/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Humans , Transcription Factor TFIIB/genetics , Transcription Factor TFIIB/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Transcription Factors, TFII/genetics , Transcription, Genetic
9.
Biol Pharm Bull ; 38(2): 263-9, 2015.
Article in English | MEDLINE | ID: mdl-25747985

ABSTRACT

Acyl CoA:diacylglycerol acyltransferase (DGAT) 1 is an enzyme that catalyzes the final step in triglyceride (TG) synthesis. This enzyme is considered to be a potential therapeutic target for obesity and diabetes. Here, results of an investigation of the pharmacological effects of JTT-553 [trans-5'-(4-amino-7,7-dimethyl-2-trifluoromethyl-7H-pyrimido[4,5-b][1,4]oxazin-6-yl)-2',3'-dihydrospiro(cyclohexane-1,1'-inden)-4-yl]acetic acid monobenzenesulfonate, a novel DGAT1 inhibitor, are reported. To measure the inhibitory activity of JTT-553 against DGAT1, TG synthesis using [(14)C]-labeled oleoyl-CoA was evaluated. Similarly, the inhibitory activity of JTT-553 against DGAT2, an isozyme of DGAT1, and acyl-CoA cholesterol acyltransferase (ACAT) 1, which is highly homologous to DGAT1, were evaluated. JTT-553 selectively inhibited human DGAT1 and showed comparable inhibitory effects on the activity of human, rat, and mouse DGAT. In vivo, JTT-553 suppressed plasma TG and chylomicron TG levels after olive oil loading in Sprague-Dawley (SD) rats. JTT-553 also inhibited TG synthesis in epididymal fat after [(14)C] oleic acid injection in C57BL/6J mice. Food intake was evaluated in SD rats fed 3.1%, 13%, or 35% (w/w) fat diets. In rats fed the 35% fat diet, JTT-553 reduced food intake. This reduction of food intake was observed 2 h after feeding, lasted for 24 h, and correlated with dietary fat content. Furthermore, JTT-553 reduced daily food intake and body weight gain in diet-induced obese rats after 4-week repeated administration. JTT-553 exerted multiple effects on intestinal fat absorption, adipose fat synthesis, and food intake, and consequently induced body weight reduction. Therefore, JTT-553 is expected to be an effective novel therapeutic agent for the treatment of obesity.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Oxazines/pharmacology , Spiro Compounds/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Body Weight/drug effects , Diacylglycerol O-Acyltransferase/metabolism , Eating , Enzyme Inhibitors/therapeutic use , Humans , Intestine, Small/drug effects , Intestine, Small/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Obesity/drug therapy , Oxazines/therapeutic use , Rats , Rats, Sprague-Dawley , Spiro Compounds/therapeutic use , Triglycerides/blood , Triglycerides/metabolism
10.
J Biol Chem ; 288(29): 20955-20965, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23749998

ABSTRACT

The Mediator complex (Mediator) plays pivotal roles in activating transcription by RNA polymerase II, but relatively little is known about its roles in repression. Here, we identified the histone arginine methyltransferase PRMT5 and WD repeat protein 77/methylosome protein 50 (WDR77/MEP50) as Mediator cyclin-dependent kinase (CDK)-interacting proteins and studied the roles of PRMT5 in the transcriptional regulation of CCAAT enhancer-binding protein (C/EBP) ß target genes. First, we purified CDK8- and CDK19-containing complexes from HeLa nuclear extracts and subjected these purified complexes to mass spectrometric analyses. These experiments revealed that two Mediator CDKs, CDK8 and CDK19, individually interact with PRMT5 and WDR77, and their interactions with PRMT5 cause transcriptional repression of C/EBPß target genes by regulating symmetric dimethylation of histone H4 arginine 3 (H4R3me2s) in the promoter regions of those genes. Furthermore, the recruitment of the DNA methyltransferase DNMT3A correlated with H4R3 dimethylation potentially leading to DNA methylation at the promoter proximal region and tight inhibition of preinitiation complex formation. In vertebrates, C/EBPß regulates many genes involved in immune responses and cell differentiation. These findings shed light on the molecular mechanisms of the repressive roles of Mediator CDKs in transcription of C/EBPß target genes and might provide clues that enable future studies of the functional associations between Mediators and epigenetic regulation.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Epigenesis, Genetic , Immunity/genetics , Mediator Complex/metabolism , Transcription, Genetic , Arginine/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Extracts , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Gene Knockdown Techniques , HeLa Cells , Histones/metabolism , Humans , Methylation , Models, Biological , Promoter Regions, Genetic , Protein Binding/genetics , Protein Subunits/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation
11.
EMBO J ; 27(7): 1161-71, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18354501

ABSTRACT

RNA polymerase II and general transcription factors (GTFs) assemble on a promoter to form a transcription preinitiation complex (PIC). Among the GTFs, TFIIE recruits TFIIH to complete the PIC formation and regulates enzymatic activities of TFIIH. However, the mode of binding between TFIIE and TFIIH is poorly understood. Here, we demonstrate the specific binding of the C-terminal acidic domain (AC-D) of the human TFIIEalpha subunit to the pleckstrin homology domain (PH-D) of the human TFIIH p62 subunit and describe the solution structures of the free and PH-D-bound forms of AC-D. Although the flexible N-terminal acidic tail from AC-D wraps around PH-D, the core domain of AC-D also interacts with PH-D. AC-D employs an entirely novel binding mode, which differs from the amphipathic helix method used by many transcriptional activators. So the binding surface between PH-D and AC-D is much broader than the specific binding surface between PH-D and the p53 acidic fragments. From our in vitro studies, we demonstrate that this interaction could be a switch to replace p53 with TFIIE on TFIIH in transcription.


Subject(s)
Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/metabolism , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Binding Sites , Humans , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Trans-Activators/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism , Transcription, Genetic
12.
Genes Cells ; 16(12): 1208-18, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22117896

ABSTRACT

Mediator is a large complex containing up to 30 subunits that consist of four modules each: head, middle, tail and CDK/Cyclin. Recent studies have shown that CDK8, a subunit of the CDK/Cyclin module, is one of the key subunits of Mediator that mediates its pivotal roles in transcriptional regulation. In addition to CDK8, CDK19 was identified in human Mediator with a great deal of similarity to CDK8 but was conserved only in vertebrates. Previously, we reported that human CDK19 could form the Mediator complexes independent of CDK8. To further investigate the in vivo transcriptional activities of the complexes, we used a luciferase assay in combined with siRNA-mediated knockdown to show that CDK8 and CDK19 possess opposing functions in viral activator VP16-dependent transcriptional regulation. CDK8 supported transcriptional activation, whereas CDK19, however, counteracted it. In this study, we further characterized CDK19. We used microarrays to identify target genes for each CDK, and we selected six genes: two target genes of CDK8, two target genes of CDK19 and two genes that were targets for both. Surprisingly, it turned out that both CDKs bound to all six target genes, regardless of their effects in transcription upon binding, suggesting Mediator as a context-specific transcriptional regulator.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation , Mediator Complex/metabolism , Cell Line, Tumor , HeLa Cells , Humans , Organ Specificity , Phosphorylation , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcriptome
13.
J Biochem ; 170(2): 203-213, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-33982754

ABSTRACT

N 6-methyladenosine (m6A), the most abundant modification in eukaryotic mRNAs, plays an important role in mRNA metabolism and functions. When adenosine is transcribed as the first cap-adjacent nucleotide, it is methylated at the ribose 2'-O and N6 positions, thus generating N6, 2'-O-dimethyladenosine (m6Am). Phosphorylated C-terminal domain (CTD)-interacting factor 1 (PCIF1) is a novel cap-specific adenine N6-methyltransferase responsible for m6Am formation. As PCIF1 specifically interacts with the Ser5-phosphorylated CTD of RNA polymerase II (Pol II), which is a marker for the early phase of transcription, PCIF1 is speculated to be recruited to the early elongating Pol II. In this study, subcellular fractionation and immunofluorescence microscopy demonstrated that PCIF1 is mainly localized to the transcriptionally active chromatin regions in HeLa cells. Chromatin immunoprecipitation (ChIP) revealed that PCIF1 was predominantly localized to the promoter of a broad range of Pol II-transcribed genes, including several protein-coding genes and non-coding RNA genes. Moreover, PCIF1 accumulation on these promoters depended entirely on transcriptional activity and Ser5 phosphorylation of the CTD. These results suggest that PCIF1 dynamically localizes to the Pol II early in transcription and may efficiently catalyze N6-methylation of the first adenosine residue of nascent mRNAs cotranscriptionally.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine/analogs & derivatives , Methyltransferases/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenosine/genetics , Adenosine/metabolism , Chromatin/metabolism , HeLa Cells , Humans , Methylation , Methyltransferases/genetics , Nuclear Proteins/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Transport , RNA Polymerase II/metabolism , RNA, Messenger/genetics , Transcription, Genetic
14.
Genes Cells ; 14(3): 395-405, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19210545

ABSTRACT

The human general transcription factor, TFIIE, consists of two subunits, alpha and beta. Structural analyses indicated the presence of a forkhead motif within the central region of TFIIEbeta. This motif was essential for transcription and possessed a double-stranded DNA-binding activity. Protein-DNA photo-cross-linking studies indicated that TFIIEbeta binds within the promoter region, adjacent to the transcription initiation site where promoter melting begins at transcription initiation. Furthermore, neither TFIIE nor the other general transcription factor TFIIH, were required for basal transcription of adenovirus major late promoter artificially pre-melted at the initiation site. These data suggest a model in which TFIIE binds to a position adjacent to the initiation site via the forkhead domain, enabling TFIIH to begin opening the promoter. Here, we used systematic point mutations to further investigate the functional roles of this domain. The mutant proteins were expressed in bacteria, purified and used to examine transcription of two different forms of template, phosphorylation of the C-terminal domain of RNA polymerase II, as well as dsDNA-binding. Taken together, our results strongly demonstrated that the primary function of the forkhead region is dsDNA-binding in transcription. In addition, we identified three positively charged lysine residues which play a key role in this function.


Subject(s)
DNA/metabolism , Transcription Factors, TFII/metabolism , Transcription, Genetic , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Phosphorylation , Point Mutation , Promoter Regions, Genetic , Protein Structure, Tertiary , Transcription Factor TFIIH/metabolism , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/genetics
15.
Genes Cells ; 14(9): 1105-18, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19682092

ABSTRACT

The carboxy-terminal domain (CTD) of the RNA polymerase II (Pol II) largest subunit undergoes reversible phosphorylation during transcription cycle. The phosphorylated CTD plays critical roles in coordinating transcription with chromatin modification and RNA processing by serving as a scaffold to recruit various proteins. Recently, we identified a novel human WW domain-containing protein PCIF1 as a phosphorylated CTD-interacting factor and demonstrated that PCIF1 negatively modulates Pol II activity in vivo. In the present study, to explore cellular functions of PCIF1, we generated PCIF1-deficient chicken DT40 cell lines. We observed significant up-regulation of WW domain-containing prolyl isomerase Pin1 in two independently established PCIF1-deficient mutant clones. As reconstitution of PCIF1 in the mutants did not reduce Pin1 expression, PCIF1 may not be a negative regulator of Pin1 expression. We assume that Pin1 over-expression might suppress defects caused by PCIF1 deficiency in DT40 cells. We furthermore compared PCIF1 and Pin1 for their functional properties and found that these two proteins exhibit most similar target specificity among other CTD-binding WW proteins, overlapping subcellular localization and comparative inhibitory effects on transcriptional activation by Pol II in human cultured cells. These results suggest that Pin1 may have overlapping cellular function with PCIF1 in vertebrate cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/metabolism , Nuclear Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , RNA Polymerase II/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Chickens , Gene Expression Regulation , HeLa Cells/metabolism , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Nuclear Proteins/genetics , Peptidylprolyl Isomerase/genetics , Phosphorylation , RNA Polymerase II/chemistry , Transfection
16.
Genes Cells ; 13(8): 817-26, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18651850

ABSTRACT

Mediator is an essential transcriptional cofactor of RNA polymerase II (Pol II) in eukaryotes. This cofactor is a large complex containing up to 30 subunits and consisting of four modules: head, middle, tail, and CDK/Cyclin. Generally, Mediator connects transcriptional regulators, cofactors, chromatin regulators, and chromatin remodellers, with the pre-initiation complex to provide a platform for the assembly of these factors. Many previous studies have revealed that CDK8, a subunit of the CDK/Cyclin module, is one of the key subunits mediating the pivotal roles of Mediator in transcriptional regulation. In addition to CDK8, CDK11 is conserved among vertebrates as a Mediator subunit and closely resembles CDK8. While the role of CDK8 has been studied extensively, little is known of the role of CDK11 in Mediator. We purified human CDK11 (hCDK11)-containing protein complexes from an epitope-tagged hCDK11-expressing HeLa cell line and found that hCDK11 could independently form Mediator complexes devoid of human CDK8 (hCDK8). To investigate the in vivo transcriptional activity of the complex, we employed a luciferase assay. Although hCDK11 has nearly 80% amino acid sequence identity to hCDK8, siRNA-knockdown study revealed that hCDK8 and hCDK11 possess opposing functions in viral activator VP16-dependent transcriptional regulation.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Herpes Simplex Virus Protein Vmw65/genetics , Amino Acid Sequence , Animals , Cyclin-Dependent Kinase 8 , Cyclin-Dependent Kinases/chemistry , HeLa Cells , Humans , Molecular Sequence Data
17.
Protein Sci ; 17(3): 389-400, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18218714

ABSTRACT

Human general transcription factor IIF (TFIIF), a component of the transcription pre-initiation complex (PIC) associated with RNA polymerase II (Pol II), was characterized by size-exclusion chromatography (SEC), electrospray ionization mass spectrometry (ESI-MS), and chemical cross-linking. Recombinant TFIIF, composed of an equimolar ratio of alpha and beta subunits, was bacterially expressed, purified to homogeneity, and found to have a transcription activity similar to a natural one in the human in vitro transcription system. SEC of purified TFIIF, as previously reported, suggested that this protein has a size >200 kDa. In contrast, ESI-MS of the purified sample gave a molecular size of 87 kDa, indicating that TFIIF is an alphabeta heterodimer, which was confirmed by matrix-assisted laser desorption/ionization (MALDI) MS of the cross-linked TFIIF components. Recent electron microscopy (EM) and photo-cross-linking studies showed that the yeast TFIIF homolog containing Tfg1 and Tfg2, corresponding to the human alpha and beta subunits, exists as a heterodimer in the PIC, so the human TFIIF is also likely to exist as a heterodimer even in the PIC. In the yeast PIC, EM and photo-cross-linking studies showed different results for the mutual location of TFIIE and TFIIF along DNA. We have examined the direct interaction between human TFIIF and TFIIE by ESI-MS, SEC, and chemical cross-linking; however, no direct interaction was observed, at least in solution. This is consistent with the previous photo-cross-linking observation that TFIIF and TFIIE flank DNA separately on both sides of the Pol II central cleft in the yeast PIC.


Subject(s)
Transcription Factors, TFII/chemistry , Chromatography, Gel , Cross-Linking Reagents , Dimerization , Humans , Recombinant Proteins/chemistry , Solutions , Spectrometry, Mass, Electrospray Ionization , Transcription Factors, TFII/genetics , Transcription Factors, TFII/isolation & purification
18.
Biochem Biophys Res Commun ; 369(2): 449-55, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18294453

ABSTRACT

Phosphorylation of the C-terminal domain (CTD) of the largest subunit of RNA polymerase II (Pol II) regulates transcription cycle and coordinates recruitment of RNA processing factors and chromatin regulators. Recently, we reported the identification of human PCIF1 as a novel protein that directly binds to the phosphorylated CTD via its WW domain, which is highly homologous to the WW domain of human peptidylprolyl isomerase Pin1. Although PCIF1 has been shown to associate with phosphorylated Pol II, functional consequence of the interaction remains unclear. Here we further characterized the cytological, structural, and functional properties of human PCIF1. Immunofluorescence microscopy revealed that endogenous PCIF1 was colocalized with the phosphorylated Pol II and the transcription elongation factor DSIF in the cell nucleus. We also found that PCIF1 WW domain inhibits the CTD phosphatase activity of SCP1 in vitro. By examining the effect of either PCIF1 overexpression or knockdown on the transactivation of reporter gene expression by various transcriptional activation domains, we found that PCIF1 significantly repressed the transactivation depend on its CTD binding ability. These data suggest that PCIF1 modulates phosphorylation status of the CTD and negatively regulates gene expression by Pol II.


Subject(s)
Gene Expression Regulation/physiology , Kidney/metabolism , Nuclear Proteins/metabolism , RNA Polymerase II/metabolism , Adaptor Proteins, Signal Transducing , Cell Line , Humans , Phosphorylation , Protein Structure, Tertiary
19.
J Biochem ; 141(5): 601-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17405796

ABSTRACT

RNA polymerase II (Pol II) is the only polymerase to possess heptapeptide repeats in the C-terminal domain (CTD) of its large subunit. During transcription, CTD phopshorylation occurs and is maintained from initiation to termination. To date, among the three known CTD kinases possessing CDK-cyclin pairs, TFIIH is the only one that forms a preinitiation complex. The Mediator complex plays essential roles in transcription initiation and during the transition from initiation to elongation by transmitting signals from transcriptional activators to Pol II. P-TEFb specifically plays a role in transcription elongation. TFIIH and mediator phosphorylate serine 5 (Ser5) of the CTD heptapeptide repeat sequence, whereas P-TEFb phosphorylates serine 2 (Ser2). Recently, it has become clear that CTD phosphorylation is not only essential for transcription, but also as a platform for RNA processing and chromatin regulation. In this review, we discuss the central role of Pol II phosphorylation in these nuclear events.


Subject(s)
RNA Polymerase II/metabolism , Transcription, Genetic/physiology , Adaptor Proteins, Signal Transducing , Humans , Models, Biological , Models, Molecular , Nuclear Proteins/physiology , Nucleotidyltransferases/metabolism , Phosphorylation , Protein Conformation/drug effects , Protein Structure, Tertiary/physiology , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA Splicing/physiology
20.
Mol Cell Biol ; 23(8): 2914-26, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12665589

ABSTRACT

The general transcription factor TFIIE plays essential roles in both transcription initiation and the transition from initiation to elongation. Previously, we systematically deleted the structural motifs and characteristic sequences of the small subunit of human TFIIE (hTFIIE beta) to map its functional regions. Here we introduced point mutations into two regions located near the carboxy terminus of hTFIIE beta and identified the functionally essential amino acid residues that bind to RNA polymerase II (Pol II), the general transcription factors, and single-stranded DNA. Although most residues identified were essential for transcription initiation, use of an in vitro transcription assay with a linearized template revealed that several residues in the carboxy-terminal helix-loop region are crucially involved in the transition stage. Mutations in these residues also affected the ability of hTFIIE beta to stimulate TFIIH-mediated phosphorylation of the carboxy-terminal heptapeptide repeats of the largest subunit of Pol II. Furthermore, these mutations conspicuously augmented the binding of hTFIIE beta to the p44 subunit of TFIIH. The antibody study indicated that they thus altered the conformation of one side of TFIIH, consisting of p44, XPD, and Cdk-activating kinase subunits, that is essential for the transition stage. This is an important clue for elucidating the molecular mechanisms involved in the transition stage.


Subject(s)
RNA Polymerase II/metabolism , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Conserved Sequence , DNA, Complementary/genetics , HeLa Cells , Helix-Loop-Helix Motifs , Humans , In Vitro Techniques , Molecular Sequence Data , Molecular Structure , Phosphorylation , Point Mutation , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Transcription Factor TFIIH , Transcription Factors, TFII/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL